4.8 Article

Pathological mitophagy disrupts mitochondrial homeostasis in Leber's hereditary optic neuropathy

Journal

CELL REPORTS
Volume 40, Issue 3, Pages -

Publisher

CELL PRESS
DOI: 10.1016/j.celrep.2022.111124

Keywords

-

Categories

Funding

  1. Italian Association for Cancer Research [IG-23670, IG-19803]
  2. A-ROSE (Associazione Ricerca Oncologica Sperimentale Estense)
  3. Progetti di Rilevante Interesse Nazionale grants [PRIN2017E5L5P3, PRIN20177E9EPY]
  4. Italian Ministry of Health [GR-2013-02356747]
  5. European Research Council [853057-InflaPML]
  6. University of Ferrara
  7. Fondazione Umberto Veronesi
  8. Polish National Science Centre [UMO-2014/15/B/NZ1/00490]
  9. Associazione Luigi Comini Onlus, Italy
  10. Mitocon - Italy [2018-01]
  11. REORION project of the Italian Ministry of Health [RF-2018-12366703]
  12. International Foundation for Optic Nerve Diseases (IFOND)

Ask authors/readers for more resources

Patients with Leber's hereditary optic neuropathy (LHON) exhibit sustained abnormal autophagy and compartment-specific mitophagy activity, disrupting mitochondrial homeostasis and leading to defective bioenergetics and excessive reactive oxygen species production. Modulating autophagy and mitochondrial biogenesis can counteract this pathological mechanism.
Leber's hereditary optic neuropathy (LHON), a disease associated with a mitochondrial DNA mutation, is characterized by blindness due to degeneration of retinal ganglion cells (RGCs) and their axons, which form the optic nerve. We show that a sustained pathological autophagy and compartment-specific mitoph-agy activity affects LHON patient-derived cells and cybrids, as well as induced pluripotent-stem-cell-derived neurons. This is variably counterbalanced by compensatory mitobiogenesis. The aberrant quality control dis-rupts mitochondrial homeostasis as reflected by defective bioenergetics and excessive reactive oxygen spe-cies production, a stress phenotype that ultimately challenges cell viability by increasing the rate of apoptosis. We counteract this pathological mechanism by using autophagy regulators (clozapine and chlo-roquine) and redox modulators (idebenone), as well as genetically activating mitochondrial biogenesis (PGC1-a overexpression). This study substantially advances our understanding of LHON pathophysiology, providing an integrated paradigm for pathogenesis of mitochondrial diseases and druggable targets for therapy.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available